Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Article in English | MEDLINE | ID: mdl-39206543

ABSTRACT

BACKGROUND: The ANGPT (angiopoietin)-TEK (tyrosine kinase, endothelial) vascular signaling pathway plays a key role in the formation of Schlemm canal, and loss-of-function mutations in the TEK or ANGPT1 gene are associated with primary congenital glaucoma in children. In genome-wide association studies, an association was identified between protection from primary open-angle glaucoma and the single-nucleotide polymorphism rs76020419 (G>T), located within a predicted miR-145-binding site in the 3' untranslated region of ANGPT2. To date, the functional impact of this variant in the anterior chamber of the eye remains largely unexplored. METHODS: MT (mutant) mice harboring an orthologous rs76020419 minor allele (T) were generated using CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9). Plasma and tissue samples, including eyes, were collected, and ANGPT2 expression was quantified using ELISA. Anterior segments from eyes were collected from WT (wild-type) and MT mice, and Schlemm canal area was quantified. RESULTS: In the MT group, higher ANGPT2 concentrations were observed in the plasma, lungs, kidneys, and eyes (P=0.0212, P<0.001, P=0.0815, and P=0.0215, respectively). Additionally, the Schlemm canal was larger in MT mice compared with WT mice (P=0.0430). CONCLUSIONS: The rs76020419 minor allele (T) is associated with increased levels of ANGPT2 and a larger Schlemm canal in mice. These findings suggest a potential protective mechanism in glaucoma.

2.
J Am Soc Nephrol ; 34(6): 969-987, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36787763

ABSTRACT

SIGNIFICANCE STATEMENT: Ischemia-reperfusion AKI (IR-AKI) is common and causes significant morbidity. Effective treatments are lacking. However, preclinical studies suggest that inhibition of angiopoietin-Tie2 vascular signaling promotes injury, whereas activation of Tie2 is protective. We show that kidney ischemia leads to increased levels of the endothelial-specific phosphatase vascular endothelial protein tyrosine phosphatase (VE-PTP; PTPRB), which inactivates Tie2. Activation of Tie2 through VE-PTP deletion, or delivery of a novel angiopoietin mimetic (Hepta-ANG1), abrogated IR-AKI in mice. Single-cell RNAseq analysis showed Tie2 activation promotes increased Entpd1 expression, downregulation of FOXO1 target genes in the kidney vasculature, and emergence of a new subpopulation of glomerular endothelial cells. Our data provide a molecular basis and identify a candidate therapeutic to improve endothelial integrity and kidney function after IR-AKI. BACKGROUND: Ischemia-reperfusion AKI (IR-AKI) is estimated to affect 2%-7% of all hospitalized patients. The significant morbidity and mortality associated with AKI indicates urgent need for effective treatments. Previous studies have shown activation of the vascular angiopoietin-Tie2 tyrosine kinase signaling pathway abrogates ischemia-reperfusion injury (IRI). We extended previous studies to (1) determine the molecular mechanism(s) underlying kidney injury and protection related to decreased or increased activation of Tie2, respectively, and (2) to test the hypothesis that deletion of the Tie2 inhibitory phosphatase vascular endothelial protein tyrosine phosphatase (VE-PTP) or injection of a new angiopoietin mimetic protects the kidney from IRI by common molecular mechanism(s). METHODS: Bilateral IR-AKI was performed in VE-PTP wild-type or knockout mice and in C57BL/6J mice treated with Hepta-ANG1 or vehicle. Histologic, immunostaining, and single-cell RNA sequencing analyses were performed. RESULTS: The phosphatase VE-PTP, which negatively regulates the angiopoietin-Tie2 pathway, was upregulated in kidney endothelial cells after IRI, and genetic deletion of VE-PTP in mice protected the kidney from IR-AKI. Injection of Hepta-ANG1 potently activated Tie2 and protected the mouse kidney from IRI. Single-cell RNAseq analysis of kidneys from Hepta-ANG1-treated and vehicle-treated mice identified endothelial-specific gene signatures and emergence of a new glomerular endothelial subpopulation associated with improved kidney function. Overlap was found between endothelial-specific genes upregulated by Hepta-ANG1 treatment and those downregulated in HUVECs with constitutive FOXO1 activation, including Entpd1 / ENTPD1 that modulates purinergic receptor signaling. CONCLUSIONS: Our data support a key role of the endothelium in the development of IR-AKI, introduce Hepta-ANG1 as a putative new therapeutic biologic, and report a model to explain how IRI reduces Tie2 signaling and how Tie2 activation protects the kidney. PODCAST: This article contains a podcast at https://dts.podtrac.com/redirect.mp3/www.asn-online.org/media/podcast/JASN/2023_05_23_JSN_Ang_EP23_052323.mp3.


Subject(s)
Acute Kidney Injury , Endothelial Cells , Mice , Animals , Endothelial Cells/metabolism , Angiopoietins/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Mice, Inbred C57BL , Endothelium/metabolism , Kidney/metabolism , Signal Transduction , Receptor, TIE-2/genetics , Angiopoietin-1/therapeutic use , Mice, Knockout , Acute Kidney Injury/prevention & control , Acute Kidney Injury/metabolism , Ischemia/complications , Ischemia/metabolism
3.
Am J Physiol Renal Physiol ; 324(1): F91-F105, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36395385

ABSTRACT

Microvascular dysfunction is a key driver of kidney disease. Pathophysiological changes in the kidney vasculature are regulated by vascular endothelial growth factor receptors (VEGFRs), supporting them as potential therapeutic targets. The tyrosine kinase receptor VEGFR-3, encoded by FLT4 and activated by the ligands VEGF-C and VEGF-D, is best known for its role in lymphangiogenesis. Therapeutically targeting VEGFR-3 to modulate lymphangiogenesis has been proposed as a strategy to treat kidney disease. However, outside the lymphatics, VEGFR-3 is also expressed in blood vascular endothelial cells in several tissues including the kidney. Here, we show that Vegfr-3 is expressed in fenestrated microvascular beds within the developing and adult mouse kidney, which include the glomerular capillary loops. We found that expression levels of VEGFR-3 are dynamic during glomerular capillary loop development, with the highest expression observed during endothelial cell migration into the S-shaped glomerular body. We developed a conditional knockout mouse model for Vegfr-3 and found that loss of Vegfr-3 resulted in a striking glomerular phenotype characterized by aneurysmal dilation of capillary loops, absence of mesangial structure, abnormal interendothelial cell junctions, and poor attachment between glomerular endothelial cells and the basement membrane. In addition, we demonstrated that expression of the VEGFR-3 ligand VEGF-C by podocytes and mesangial cells is dispensable for glomerular development. Instead, VEGFR-3 in glomerular endothelial cells attenuates VEGFR-2 phosphorylation. Together, the results of our study support a VEGF-C-independent functional role for VEGFR-3 in the kidney microvasculature outside of lymphatic vessels, which has implications for clinical therapies that target this receptor.NEW & NOTEWORTHY Targeting VEGFR-3 in kidney lymphatics has been proposed as a method to treat kidney disease. However, expression of VEGFR-3 is not lymphatic-specific. We demonstrated developmental expression of VEGFR-3 in glomerular endothelial cells, with loss of Vegfr-3 leading to malformation of glomerular capillary loops. Furthermore, we showed that VEGFR-3 attenuates VEGFR-2 activity in glomerular endothelial cells independent of paracrine VEGF-C signaling. Together, these data provide valuable information for therapeutic development targeting these pathways.


Subject(s)
Kidney Diseases , Vascular Endothelial Growth Factor Receptor-3 , Mice , Animals , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism , Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Capillaries/metabolism
4.
FASEB J ; 33(8): 9602-9615, 2019 08.
Article in English | MEDLINE | ID: mdl-31150280

ABSTRACT

Obesity is a leading cause of chronic kidney disease (CKD), but how obesity promotes renal injury remains poorly understood. Here we showed that ATP-citrate lyase (ACL), an enzyme converting citrate to acetyl-CoA, is highly induced in the kidney of overweight or obese patients with CKD and ob/ob BTBR mice. ACL induction is associated with increased ectopic lipid accumulation (ELA), glomerulosclerosis, and albuminuria. Acetyl-CoA is the substrate for de novo lipogenesis as well as for histone acetylation. By raising acetyl-CoA concentration ACL promotes H3K9/14 and H3K27 hyperacetylation leading to up-regulation of several rate-limiting lipogenic enzymes and fibrogenic factors. On the other hand, the excess acetyl-CoA generated as a result of ACL induction provides the substrate for these lipogenic enzymes to drive de novo lipogenesis leading to ELA, a detrimental event toward renal injury. In mesangial cells, ACL is synergistically induced by high glucose, palmitate, and TNF-α via NF-κB and PKA pathways. Under these conditions, H3K9/14 and H3K27 hyperacetylation, as well as the induction of the lipogenic and fibrogenic proteins, are completely blocked in the presence of an ACL inhibitor. Collectively, these data suggest that ACL is an epigenetic regulator that promotes renal ELA and fibrogenesis leading to renal injury in obesity.-Chen, Y., Deb, D. K., Fu, X., Yi, B., Liang, Y., Du, J., He, L., Li, Y. C. ATP-citrate lyase is an epigenetic regulator to promote obesity-related kidney injury.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Epigenesis, Genetic/genetics , Obesity/enzymology , Obesity/genetics , ATP Citrate (pro-S)-Lyase/genetics , Acetyl Coenzyme A/metabolism , Acetylation/drug effects , Animals , Blotting, Western , Epigenesis, Genetic/drug effects , Female , Glucose/pharmacology , Humans , Immunohistochemistry , Male , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mice , NF-kappa B/pharmacology , Palmitic Acid/pharmacology
5.
FASEB J ; 31(5): 2065-2075, 2017 05.
Article in English | MEDLINE | ID: mdl-28148567

ABSTRACT

Hyperglycemia is a major pathogenic factor that promotes diabetic nephropathy, but the underlying mechanism remains incompletely understood. Here, we show that high glucose induced cAMP response element-binding protein (CREB)-binding protein (CBP)-mediated H3K9/14 hyperacetylation in approximately 5000 gene promoters in glomerular mesangial cells, including those of Tgfb1, Tgfb3, and Ctgf, the major profibrotic factors that are known to drive diabetic renal fibrogenesis. In these promoters, H3K9/14 hyperacetylation was closely associated with NF-κB or CREB motifs. Chromatin immunoprecipitation assays confirmed that hyperglycemia promoted phospho-p65 or phospho-CREB and CBP bindings and RNA polymerase II recruitment to these promoters in mesangial cells as well as in glomeruli that were purified from type I and type II diabetic mice. Under hyperglycemia, cAMP production and PKA activity were markedly increased as a result of glucose transporter 1-mediated glucose influx that drives glucose metabolism and ATP production, which led to increased phosphorylation of p65 and CREB. Inhibition of adenylyl cyclase or PKA activity blocked p65 and CREB phosphorylation, CBP recruitment, and histone acetylation in these promoters. Collectively, these data demonstrate that the cAMP-PKA pathway plays a key role in epigenetic regulation of key profibrotic factors in diabetes.-Deb, D. K., Bao, R., Li, Y. C. Critical role of the cAMP-PKA pathway in hyperglycemia-induced epigenetic activation of fibrogenic program in the kidney.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Epigenesis, Genetic/genetics , Hyperglycemia/metabolism , Kidney/metabolism , Signal Transduction , Animals , Diabetes Mellitus, Experimental/metabolism , Mice , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Promoter Regions, Genetic/genetics
6.
Am J Physiol Renal Physiol ; 313(2): F423-F429, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28490526

ABSTRACT

The goal of this study was to address the role of ATP-citrate lyase (ACL), an enzyme that converts citrate to acetyl-CoA, in high glucose (HG)-induced histone acetylation and profibrotic gene expression. Our recent ChIP-Seq studies have demonstrated that HG induces genome-wide histone hyperacetylation in mesangial cells (MCs). Here, we showed that exposure of MCs to HG markedly increased histone acetylation at the H3K9/14 and H3K18 marks and induced the expression of potent profibrotic factors TGF-ß1, TGF-ß3, and connective tissue growth factor (CTGF). The induction of these profibrotic factors was further enhanced by histone deacetylase inhibitor but suppressed by histone acetyl-transferase inhibitor, confirming the importance of histone acetylation in this regulation. Interestingly, HG not only upregulated ACL expression but also promoted ACL nuclear translocation, evidenced by increased ACL concentration and activity in the nuclear extracts. Consistent with this observation, transfection of MCs with a plasmid-carrying green fluorescent protein (GFP)-ACL fusion protein led to GFP nuclear accumulation when cultured in HG condition. Silencing ACL with siRNAs alleviated HG-induced histone hyperacetylation, as well as upregulation of TGF-ß1, TGF-ß3, CTGF, and extracellular matrix (ECM) proteins fibronectin and collagen type IV, whereas ACL overexpression further enhanced HG induction of histone acetylation, as well as these profibrotic factors and ECM proteins. Collectively, these observations demonstrate that HG promotes ACL expression and translocation into the nucleus, where ACL converts citrate to acetyl-CoA to provide the substrate for histone acetylation, leading to upregulation of fibrogenic genes. Therefore, ACL plays a critical role in epigenetic regulation of diabetic renal fibrosis.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Diabetic Nephropathies/enzymology , Epigenesis, Genetic/drug effects , Glucose/toxicity , Histones/genetics , Mesangial Cells/drug effects , Protein Processing, Post-Translational/drug effects , ATP Citrate (pro-S)-Lyase/genetics , Acetyl Coenzyme A/metabolism , Acetylation , Active Transport, Cell Nucleus , Animals , Cell Line, Transformed , Citric Acid/metabolism , Collagen Type IV/genetics , Collagen Type IV/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Fibronectins/genetics , Fibronectins/metabolism , Fibrosis , Mesangial Cells/enzymology , Mesangial Cells/pathology , Mice , RNA Interference , Time Factors , Transfection , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/metabolism , Up-Regulation
7.
J Immunol ; 190(7): 3687-95, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23436936

ABSTRACT

The negative feedback mechanism is essential to maintain effective immunity and tissue homeostasis. 1,25-dihydroxyvitamin D (1,25[OH]2D3) modulates innate immune response, but the mechanism remains poorly understood. In this article, we report that vitamin D receptor signaling attenuates TLR-mediated inflammation by enhancing the negative feedback inhibition. Vitamin D receptor inactivation leads to hyperinflammatory response in mice and macrophage cultures when challenged with LPS, because of microRNA-155 (miR-155) overproduction that excessively suppresses suppressor of cytokine signaling 1, a key regulator that enhances the negative feedback loop. Deletion of miR-155 attenuates vitamin D suppression of LPS-induced inflammation, confirming that 1,25(OH)2D3 stimulates suppressor of cytokine signaling 1 by downregulating miR-155. 1,25(OH)2D3 downregulates bic transcription by inhibiting NF-κB activation, which is mediated by a κB cis-DNA element located within the first intron of the bic gene. Together, these data identify a novel regulatory mechanism for vitamin D to control innate immunity.


Subject(s)
Macrophages/drug effects , Macrophages/metabolism , MicroRNAs/genetics , Signal Transduction/drug effects , Suppressor of Cytokine Signaling Proteins/genetics , Toll-Like Receptors/metabolism , Vitamin D/analogs & derivatives , Animals , Cell Line , Cytokines/immunology , Cytokines/metabolism , Enzyme Activation/drug effects , Feedback, Physiological/drug effects , Gene Expression Regulation/drug effects , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Knockout , Models, Biological , NF-kappa B/metabolism , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Suppressor of Cytokine Signaling 1 Protein , Transcription, Genetic/drug effects , Vitamin D/pharmacology
8.
J Biol Chem ; 288(27): 19450-8, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23671281

ABSTRACT

1,25-Dihydroxyvitamin D (1,25(OH)2D3) is known to suppress NF-κB activity, but the underlying mechanism remains poorly understood. Here we show that the vitamin D receptor (VDR) physically interacts with IκB kinase ß (IKKß) to block NF-κB activation. 1,25(OH)2D3 rapidly attenuates TNFα-induced p65 nuclear translocation and NF-κB activity in a VDR-dependent manner. VDR overexpression inhibits IKKß-induced NF-κB activity. GST pull-down assays and coimmunoprecipitation experiments demonstrated that VDR physically interacts with IKKß and that this interaction is enhanced by 1,25(OH)2D3. Protein mapping reveals that VDR-IKKß interaction occurs between the C-terminal portions of the VDR and IKKß proteins. Reconstitution of VDR(-/-) cells with the VDR C terminus restores the ability to block TNFα-induced NF-κB activation and IL-6 up-regulation. VDR-IKKß interaction disrupts the formation of the IKK complex and, thus, abrogates IKKß phosphorylation at Ser-177 and abolishes IKK activity to phosphorylate IκBα. Consequently, stabilization of IκBα arrests p65/p50 nuclear translocation. Together, these data define a novel mechanism whereby 1,25(OH)2D3-VDR inhibits NF-κB activation.


Subject(s)
Cell Nucleus/metabolism , I-kappa B Kinase/metabolism , NF-kappa B p50 Subunit/metabolism , Receptors, Calcitriol/metabolism , Transcription Factor RelA/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Animals , Calcitriol/pharmacology , Cell Nucleus/genetics , HEK293 Cells , Humans , I-kappa B Kinase/genetics , Mice , Mice, Knockout , NF-kappa B p50 Subunit/genetics , Peptide Mapping , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Structure, Tertiary , Receptors, Calcitriol/genetics , Transcription Factor RelA/genetics
9.
Am J Nephrol ; 38(1): 39-49, 2013.
Article in English | MEDLINE | ID: mdl-23817135

ABSTRACT

Diabetic glomerulosclerosis is characterized by accumulation of extracellular matrix proteins, mesangial expansion, and tubulointerstitial fibrosis. Hyperglycemia accelerates development of the disease, a direct result of increased intracellular glucose availability. The facilitative glucose transporter GLUT1 mediates mesangial cell glucose flux which leads to activation of signaling cascades favoring glomerulosclerosis, including pathways mediated by angiotensin II (Ang II), transforming growth factor ß (TGF-ß), connective tissue growth factor (CTGF), and vascular endothelial growth factor (VEGF). Ang II has both hemodynamic and metabolic effects directly inducing GLUT1 and/or matrix protein synthesis through diacyl glycerol (DAG) or protein kinase C (PKC) induction, mesangial cell stretch, and/or through transactivation of the epidermal growth factor receptor, the platelet-derived growth factor receptor, and the insulin-like growth factor-1 receptor, all of which may stimulate GLUT1 synthesis via an ERK-mediated pathway. Conversely, inhibition of Ang II effects suppresses GLUT1 and cellular glucose uptake. GLUT1-mediated glucose flux leads to metabolism of glucose via glycolysis, with induction of DAG, PKC, TGF-ß1, CTGF and VEGF. VEGF in turn triggers both GLUT1 and matrix synthesis. New roles for GLUT1-mTOR and GLUT1-mechano-growth factor interactions in diabetic glomerulosclerosis have also recently been suggested. Recent mouse models confirmed roles for GLUT1 in vivo in stimulating glomerular growth factor expression, growth factor receptors and development of glomerulosclerosis. GLUT1 may therefore act in concert with cytokines and growth factors to induce diabetic glomerulosclerosis. Further clarification of the pathways involved may prove useful for the therapy of diabetic nephropathy. New directions for investigation are discussed.


Subject(s)
Diabetic Nephropathies/physiopathology , Glucose Transporter Type 1/physiology , Glucose/metabolism , Hyperglycemia/physiopathology , Angiotensin II/physiology , Animals , Connective Tissue Growth Factor/physiology , Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Humans , Hyperglycemia/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Vascular Endothelial Growth Factor A/physiology
10.
J Am Soc Nephrol ; 23(12): 1977-86, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23123403

ABSTRACT

Vitamin D and its analogs have antiproteinuric activity and podocytes express the vitamin D receptor, but whether vitamin D signaling in podocytes accounts for this renoprotection is unknown. To investigate this question, we used the 2.5 kb podocin promoter to target Flag-tagged human vitamin D receptor (hVDR) to podocytes in DBA/2J mice. After the induction of diabetes with streptozotocin, transgenic mice had less albuminuria than wild-type controls. In transgenic mice, a low dose of the vitamin D analog doxercalciferol prevented albuminuria, markedly attenuated podocyte loss and apoptosis, and reduced glomerular fibrosis, but it had little effect on the progression of diabetic nephropathy in wild-type mice. Moreover, reconstitution of VDR-null mice with the hVDR transgene in podocytes rescued VDR-null mice from severe diabetes-related renal damage. In culture, 1,25-dihydroxyvitamin D suppressed high-glucose-induced apoptosis of podocytes by blocking p38- and ERK-mediated proapoptotic pathways. Taken together, these data provide strong evidence that vitamin D/VDR signaling in podocytes plays a critical role in the protection of the kidney from diabetic injury.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/metabolism , Hyperglycemia/complications , Podocytes/metabolism , Receptors, Calcitriol/metabolism , Animals , Apoptosis , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/genetics , Female , Humans , Hyperglycemia/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Inbred DBA , Mice, Knockout , Pregnancy , Promoter Regions, Genetic
11.
bioRxiv ; 2023 May 03.
Article in English | MEDLINE | ID: mdl-37205366

ABSTRACT

Background: Lymphangiogenesis is believed to be a protective response in the setting of multiple forms of kidney injury and mitigates the progression of interstitial fibrosis. To augment this protective response, promoting kidney lymphangiogenesis is being investigated as a potential treatment to slow the progression of kidney disease.As injury related lymphangiogenesis is driven by signaling from the receptor VEGFR-3 in response to the cognate growth factor VEGF-C released by tubular epithelial cells, this signaling pathway is a candidate for future kidney therapeutics. However, the consequences to kidney development and function to targeting this signaling pathway remains poorly defined. Methods: We generated a new mouse model expressing Vegf-C under regulation of the nephron progenitor Six2Cre driver strain (Six2Vegf-C). Mice underwent a detailed phenotypic evaluation. Whole kidneys were processed for histology and micro computed tomography 3-dimensional imaging. Results: Six2Vegf-C mice had reduced body weight and kidney function compared to littermate controls. Six2Vegf-C kidneys demonstrated large peripelvic fluid filled lesions with distortion of the pelvicalcyceal system which progressed in severity with age. 3D imaging showed a 3-fold increase in total cortical vascular density. Histology confirmed a substantial increase in LYVE1+/PDPN+/VEGFR3+ lymphatic capillaries extending alongside EMCN+ peritubular capillaries. There was no change in EMCN+ peritubular capillary density. Conclusions: Kidney lymphangiogenesis was robustly induced in the Six2Vegf-C mice. There were no changes in peritubular blood capillary density despite these endothelial cells also expressing VEGFR-3. The model resulted in a severe cystic kidney phenotype that resembled a human condition termed renal lymphangiectasia. This study defines the vascular consequences of augmenting VEGF-C signaling during kidney development and provides new insight into a mimicker of human cystic kidney disease.

12.
J Biol Chem ; 286(37): 32011-7, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21803771

ABSTRACT

Nephrin plays a key role in maintaining the structure of the slit diaphragm in the glomerular filtration barrier. Our previous studies have demonstrated potent renoprotective activity for 1,25-dihydroxyvitamin D (1,25(OH)(2)D(3)). Here we showed that in podocytes 1,25(OH)(2)D(3) markedly stimulated nephrin mRNA and protein expression. ChIP scan of the 6-kb 5' upstream region of the mouse nephrin gene identified several putative vitamin D response elements (VDREs), and EMSA confirmed that the VDRE at -312 (a DR4-type VDRE) could be bound by vitamin D receptor (VDR)/retinoid X receptor. Luciferase reporter assays of the proximal nephrin promoter fragment (-427 to +173) showed strong induction of luciferase activity upon 1,25(OH)(2)D(3) treatment, and the induction was abolished by mutations within -312VDRE. ChIP assays showed that, upon 1,25(OH)(2)D(3) activation, VDR bound to this VDRE leading to recruitment of DRIP205 and RNA polymerase II and histone 4 acetylation. Treatment of mice with a vitamin D analog induced nephrin mRNA and protein in the kidney, accompanied by increased VDR binding to the -312VDRE and histone 4 acetylation. 1,25(OH)(2)D(3) reversed high glucose-induced nephrin reduction in podocytes, and vitamin D analogs prevented nephrin decline in both type 1 and 2 diabetic mice. Together these data demonstrate that 1,25(OH)(2)D(3) stimulates nephrin expression in podocytes by acting on a VDRE in the proximal nephrin promoter. Nephrin up-regulation likely accounts for part of the renoprotective activity of vitamin D.


Subject(s)
Bone Density Conservation Agents/pharmacology , Calcitriol/pharmacology , Gene Expression Regulation/drug effects , Membrane Proteins/biosynthesis , Podocytes/metabolism , Vitamin D Response Element , Acetylation/drug effects , Animals , Cell Line, Transformed , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , HEK293 Cells , Histones/genetics , Histones/metabolism , Humans , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , Membrane Proteins/genetics , Mice , Mutation , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Calcitriol/agonists , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism
13.
J Biol Chem ; 286(39): 33804-10, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21840998

ABSTRACT

Our previous studies demonstrated a high fat diet-resistant lean phenotype of vitamin D receptor (VDR)-null mutant mice mainly due to increased energy expenditure, suggesting an involvement of the VDR in energy metabolism. Here, we took a transgenic approach to further define the role of VDR in adipocyte biology. We used the aP2 gene promoter to target the expression of the human (h) VDR in adipocytes in mice. In contrast to the VDR-null mice, the aP2-hVDR Tg mice developed obesity compared with the wild-type counterparts without changes in food intake. The increase in fat mass was mainly due to markedly reduced energy expenditure, which was correlated with decreased locomotive activity and reduced fatty acid ß-oxidation and lipolysis in the adipose tissue in the transgenic mice. Consistently, the expression of genes involved in the regulation of fatty acid transport, thermogenesis, and lipolysis were suppressed in the transgenic mice. Taken together, these data confirm an important role of the VDR in the regulation of energy metabolism.


Subject(s)
Adipocytes/metabolism , Energy Metabolism , Fatty Acids/metabolism , Gene Expression Regulation , Obesity/metabolism , Receptors, Calcitriol/biosynthesis , Adipocytes/physiology , Animals , Biological Transport, Active/genetics , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Humans , Lipolysis/genetics , Locomotion/genetics , Mice , Mice, Mutant Strains , Mice, Transgenic , Obesity/genetics , Obesity/pathology , Organ Specificity , Oxidation-Reduction , Promoter Regions, Genetic/genetics , Receptors, Calcitriol/genetics , Thermogenesis/genetics
14.
Int J Cancer ; 130(1): 10-9, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21328347

ABSTRACT

Emerging evidence supports an inhibitory role for vitamin D in colorectal carcinogenesis; however, the mechanism remains unclear. The adenomatous polyposis coli (APC)/ß-catenin pathway plays a critical role in colorectal carcinogenesis. The purpose of our study is to explore the interactions of vitamin D and APC/ß-catenin pathways in intestinal tumor development. APC(min/+) mice with genetic inactivation of the vitamin D receptor (VDR) were generated through breeding. Intestinal tumorigenesis was compared between APC(min/+) and APC(min/+) VDR(-/-) mice at different ages. No differences were seen in the number of small intestinal and colonic tumors between APC(min/+) and APC(min/+) VDR(-/-) mice aged 3, 4, 6 and 7 months. The size of the tumors, however, was significantly increased in APC(min/+) VDR(-/-) mice in all age groups. Immunostaining showed significant increases in ß-catenin, cyclin D1, phosphorylated Stat-3 and MSH-2 levels and decreases in Stat-1 in APC(min/+) VDR(-/-) tumors compared to APC(min/+) tumors. These observations suggest that VDR signaling inhibits tumor growth rather than tumor initiation in the intestine. Thus, the increased tumor burden in APC(min/+) VDR(-/-) mice is likely due to the loss of the growth-inhibiting effect of VDR. This study provides strong evidence for the in vivo relevance of the interaction demonstrated in vitro between the vitamin D and ß-catenin signaling pathways in intestinal tumorigenesis.


Subject(s)
Genes, APC/physiology , Intestinal Neoplasms/etiology , Intestinal Neoplasms/pathology , Receptors, Calcitriol/physiology , Animals , Blotting, Western , Immunoenzyme Techniques , Immunoprecipitation , Intestinal Neoplasms/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Receptors, Calcitriol/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , beta Catenin/metabolism
15.
Arch Biochem Biophys ; 507(2): 241-7, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21176770

ABSTRACT

Plasminogen activator inhibitor (PAI)-1 is a major fibrinolytic inhibitor. High PAI-1 is associated with increased renal and cardiovascular disease risk. Previous studies demonstrated PAI-1 down-regulation by 1,25-dihydroxyvitamin D3 (1,25(OH)2D3), but the molecular mechanism remains unknown. Here we show that exposure of mouse embryonic fibroblasts to TNFα or LPS led to a marked induction of PAI-1, which was blunted by 1,25(OH)2D3, NF-κB inhibitor or p65 siRNA, suggesting the involvement of NF-κB in 1,25(OH)2D3-induced repression. In mouse Pai-1 promoter a putative cis-κB element was identified at -299. EMSA and ChIP assays showed that TNF-α increased p50/p65 binding to this κB site, which was disrupted by 1,25(OH)2D3. Luciferase reporter assays showed that PAI-1 promoter activity was induced by TNFα or LPS, and the induction was blocked by 1,25(OH)2D3. Mutation of the κB site blunted TNFα, LPS or 1,25(OH)2D3 effects. 1,25(OH)2D3 blocked IκBα degradation and arrested p50/p65 nuclear translocation. In mice LPS stimulated PAI-1 expression in the heart and macrophages, and the stimulation was blunted by pre-treatment with a vitamin D analog. Together these data demonstrate that 1,25(OH)2D3 down-regulates PAI-1 by blocking NF-κB activation. Inhibition of PAI-1 production may contribute to the reno- and cardio-protective effects of vitamin D.


Subject(s)
Calcitriol/pharmacology , Gene Expression Regulation/drug effects , NF-kappa B/metabolism , Plasminogen Activator Inhibitor 1/genetics , Active Transport, Cell Nucleus/drug effects , Animals , Base Sequence , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Down-Regulation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/pharmacology , Mice , Promoter Regions, Genetic/genetics , Tumor Necrosis Factor-alpha/pharmacology
16.
Proc Natl Acad Sci U S A ; 105(41): 15896-901, 2008 Oct 14.
Article in English | MEDLINE | ID: mdl-18838678

ABSTRACT

The renin-angiotensin system (RAS) plays a critical role in the development of diabetic nephropathy, and blockade of the RAS is currently used for treatment of diabetic nephropathy. One major problem for the current RAS inhibitors is the compensatory renin increase, which reduces the efficacy of RAS inhibition. We have shown that vitamin D exerts renoprotective actions by transcriptionally suppressing renin. Here we demonstrated that combination therapy with an AT1 receptor blocker and a vitamin D analog markedly ameliorated renal injury in the streptozotocin (STZ)-induced diabetes model due to the blockade of the compensatory renin rise by the vitamin D analog, leading to more effective RAS inhibition. STZ-treated diabetic DBA/2J mice developed progressive albuminuria and glomerulosclerosis within 13 weeks, accompanied by increased intrarenal production of angiotensin (Ang) II, fibronection, TGF-beta, and MCP-1 and decreased expression of slit diaphragm proteins. Treatment of the diabetic mice with losartan or paricalcitol (19-nor-1,25-dihydroxyvitamin D(2), an activated vitamin D analog) alone moderately ameliorated kidney injury; however, combined treatment with losartan and paricalcitol completely prevented albuminuria, restored glomerular filtration barrier structure, and markedly reduced glomerulosclerosis. The combined treatment suppressed the induction of fibronection, TGF-beta, and MCP-1 and reversed the decline of slit diaphragm proteins nephrin, Neph-1, ZO-1, and alpha-actinin-4. These were accompanied by blockade of intrarenal renin and Ang II accumulation induced by hyperglycemia and losartan. These data demonstrate that inhibition of the RAS with combination of vitamin D analogs and RAS inhibitors effectively prevents renal injury in diabetic nephropathy.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Diabetic Nephropathies/drug therapy , Renin/drug effects , Vitamin D/therapeutic use , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Diabetes Mellitus, Experimental/drug therapy , Drug Combinations , Ergocalciferols/pharmacology , Ergocalciferols/therapeutic use , Losartan/pharmacology , Losartan/therapeutic use , Mice , Receptor, Angiotensin, Type 1 , Renin/genetics , Renin-Angiotensin System , Streptozocin , Vitamin D/analogs & derivatives
17.
J Am Soc Nephrol ; 21(6): 966-73, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20378820

ABSTRACT

Analogs of vitamin D attenuate renal injury in several models of kidney disease, but the mechanism underlying this renoprotective effect is unknown. To address the role of the vitamin D receptor (VDR) in renal fibrogenesis, we subjected VDR-null mice to unilateral ureteral obstruction for 7 days. Compared with wild-type mice, VDR-null mice developed more severe renal damage in the obstructed kidney, with marked tubular atrophy and interstitial fibrosis. Significant induction of extracellular matrix proteins (fibronectin and collagen I), profibrogenic and proinflammatory factors (TGF-beta, connective tissue growth factor, and monocyte chemoattractant protein 1), and epithelial-to-mesenchymal transition accompanied this histologic damage. Because VDR ablation activates the renin-angiotensin system and leads to accumulation of angiotensin II (AngII) in the kidney, we assessed whether elevated AngII in the VDR-null kidney promotes injury. Treatment with the angiotensin type 1 antagonist losartan eliminated the difference in obstruction-induced interstitial fibrosis between wild-type and VDR-null mice, suggesting that AngII contributes to the enhanced renal fibrosis observed in obstructed VDR-null kidneys. Taken together, these results suggest that the VDR attenuates obstructive renal injury at least in part by suppressing the renin-angiotensin system.


Subject(s)
Kidney Diseases/pathology , Kidney Diseases/prevention & control , Kidney/pathology , Receptors, Calcitriol/physiology , Renin-Angiotensin System/physiology , Angiotensin I/antagonists & inhibitors , Animals , Cells, Cultured , Chemokine CCL2/physiology , Collagen Type I/physiology , Connective Tissue Growth Factor/physiology , Disease Models, Animal , Fibronectins/physiology , Fibrosis/pathology , Fibrosis/physiopathology , Fibrosis/prevention & control , Kidney/physiopathology , Kidney Diseases/physiopathology , Losartan/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Calcitriol/genetics , Transforming Growth Factor beta/physiology
18.
Epigenetics ; 16(12): 1317-1334, 2021 12.
Article in English | MEDLINE | ID: mdl-33356812

ABSTRACT

Because ADAM17 promotes colonic tumorigenesis, we investigated potential miRNAs regulating ADAM17; and examined effects of diet and tumorigenesis on these miRNAs. We also examined pre-miRNA processing and tumour suppressor roles of several of these miRNAs in experimental colon cancer. Using TargetScan, miR-145, miR-148a, and miR-152 were predicted to regulate ADAM17. miR-143 was also investigated as miR-143 and miR-145 are co-transcribed and associated with decreased tumour growth. HCT116 colon cancer cells (CCC) were co-transfected with predicted ADAM17-regulating miRNAs and luciferase reporters controlled by ADAM17-3'UTR. Separately, pre-miR-143 processing by colonic cells was measured. miRNAs were quantified by RT-PCR. Tumours were induced with AOM/DSS in WT and transgenic mice (Tg) expressing pre-miR-143/miR-145 under villin promoter. HCT116 transfection with miR-145, -148a or -152, but not scrambled miRNA inhibited ADAM17 expression and luciferase activity. The latter was suppressed by mutations in ADAM17-3'UTR. Lysates from colonocytes, but not CCC, processed pre-miR-143 and mixing experiments suggested CCC lacked a competency factor. Colonic miR-143, miR-145, miR-148a, and miR-152 were downregulated in tumours and more moderately by feeding mice a Western diet. Tg mice were resistant to DSS colitis and had significantly lower cancer incidence and tumour multiplicity. Tg expression blocked up-regulation of putative targets of miR-143 and miR-145, including ADAM17, K-Ras, XPO5, and SET. miR-145, miR-148a, and miR-152 directly suppress colonocyte ADAM17 and are down-regulated in colon cancer. This is the first direct demonstration of tumour suppressor roles for miR-143 and miR-145 in an in vivo model of colonic tumorigenesis.


Subject(s)
Colitis , Colonic Neoplasms , MicroRNAs , Animals , Colonic Neoplasms/genetics , DNA Methylation , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Inflammation , Karyopherins , Mice , MicroRNAs/metabolism , Up-Regulation
19.
Am J Physiol Renal Physiol ; 299(1): F99-F111, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20375117

ABSTRACT

Previous work identified an important role for hyperglycemia in diabetic nephropathy (The Diabetes Control and Complications Trial Research Group. N Engl J Med 329: 977-986, 1993; UK Prospective Diabetes Study Group. Lancet 352: 837-853, 1998), and increased glomerular GLUT1 has been implicated. However, the roles of GLUT1 and intracellular glucose have not been determined. Here, we developed transgenic GLUT1-overexpressing mice (GT1S) to characterize the roles of GLUT1 and intracellular glucose in the development of glomerular disease without diabetes. GLUT1 was overexpressed in glomerular mesangial cells (MC) of C57BL6 mice, a line relatively resistant to diabetic nephropathy. Blood pressure, blood glucose, glomerular morphometry, matrix proteins, cell signaling, transcription factors, and selected growth factors were examined. Kidneys of GT1S mice overexpressed GLUT1 in glomerular MCs and small vessels, rather than renal tubules. GT1S mice were neither diabetic nor hypertensive. Glomerular GLUT1, glucose uptake, mean capillary diameter, and mean glomerular volume were all increased in the GT1S mice. Moderately severe glomerulosclerosis (GS) was established by 26 wk of age in GT1S mice, with increased glomerular type IV collagen and fibronectin. Modest increases in glomerular basement membrane thickness and albuminuria were detected with podocyte foot processes largely preserved, in the absence of podocyte GLUT1 overexpression. Activation of glomerular PKC, along with increased transforming growth factor-beta1, VEGFR1, VEGFR2, and VEGF were all detected in glomeruli of GT1S mice, likely contributing to GS. The transcription factor NF-kappaB was also activated. Overexpression of glomerular GLUT1, mimicking the diabetic GLUT1 response, produced numerous features typical of diabetic glomerular disease, without diabetes or hypertension. This suggested GLUT1 may play an important role in the development of diabetic GS.


Subject(s)
Diabetic Nephropathies/metabolism , Glucose Transporter Type 1/metabolism , Kidney Glomerulus/metabolism , Aging , Albuminuria/metabolism , Albuminuria/pathology , Animals , Blood Glucose/metabolism , Blood Pressure , Cells, Cultured , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Glomerular Basement Membrane/metabolism , Glomerular Mesangium/metabolism , Glucose Transporter Type 1/genetics , Humans , Kidney Glomerulus/pathology , Mesangial Cells/metabolism , Mesangial Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Podocytes/metabolism , Protein Kinase C/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
20.
Lab Invest ; 90(1): 83-97, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19918242

ABSTRACT

Reduced nephron numbers may predispose to renal failure. We hypothesized that glucose transporters (GLUTs) may contribute to progression of the renal disease, as GLUTs have been implicated in diabetic glomerulosclerosis and hypertensive renal disease with mesangial cell (MC) stretch. The Os (oligosyndactyly) allele that typically reduces nephron number by approximately 50%, was repeatedly backcrossed from ROP (Ra/+ (ragged), Os/+ (oligosyndactyly), and Pt/+ (pintail)) Os/+ mice more than six times into the Fvb mouse background to obtain Os/+ and +/+ mice with the Fvb background for study. Glomerular function, GLUT1, signaling, albumin excretion, and structural and ultrastructural changes were assessed. The FvbROP Os/+ mice (Fvb background) exhibited increased glomerular GLUT1, glucose uptake, VEGF, glomerular hypertrophy, hyperfiltration, extensive podocyte foot process effacement, marked albuminuria, severe extracellular matrix (ECM) protein deposition, and rapidly progressive renal failure leading to their early demise. Glomerular GLUT1 was increased 2.7-fold in the FvbROP Os/+ mice vs controls at 4 weeks of age, and glucose uptake was increased 2.7-fold. These changes were associated with the activation of glomerular PKCbeta1 and NF-kappaB p50 which contribute to ECM accumulation. The cyclic mechanical stretch of MCs in vitro, used as a model for increased MC stretch in vivo, reproduced increased GLUT1 at 48 h, a stimulus for increased VEGF expression which followed at 72 h. VEGF was also shown to act in a positive feedback manner on MC GLUT1, increasing GLUT1 expression, glucose uptake and fibronectin (FN) accumulation in vitro, whereas antisense suppression of GLUT1 largely blocked FN upregulation by VEGF. The FvbROP Os/+ mice exhibited an early increase in glomerular GLUT1 leading to increased glomerular glucose uptake PKCbeta1, and NF-kappaB activation, with excess ECM accumulation. A GLUT1-VEGF-GLUT1 positive feedback loop may play a key role in contributing to renal disease in this model of nondiabetic glomerulosclerosis.


Subject(s)
Albuminuria/etiology , Glucose Transporter Type 1/metabolism , Mice, Mutant Strains/metabolism , Nephrons/abnormalities , Renal Insufficiency/etiology , Renal Insufficiency/physiopathology , Vascular Endothelial Growth Factor A/metabolism , Alleles , Animals , Cells, Cultured , Creatinine/metabolism , Disease Progression , Extracellular Matrix Proteins/metabolism , Glomerular Mesangium/metabolism , Glomerular Mesangium/pathology , Immunohistochemistry , Isoenzymes/metabolism , Kidney/growth & development , Kidney/metabolism , Kidney/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains/genetics , Microscopy, Electron , NF-kappa B/metabolism , Protein Kinase C/metabolism , Stress, Mechanical , Syndactyly/genetics , Transforming Growth Factor beta1/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL